WO2012097269A1 - Inhibiteurs de pyrimidine gyrase et topoïsomérase iv - Google Patents

Inhibiteurs de pyrimidine gyrase et topoïsomérase iv Download PDF

Info

Publication number
WO2012097269A1
WO2012097269A1 PCT/US2012/021270 US2012021270W WO2012097269A1 WO 2012097269 A1 WO2012097269 A1 WO 2012097269A1 US 2012021270 W US2012021270 W US 2012021270W WO 2012097269 A1 WO2012097269 A1 WO 2012097269A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
infections
pharmaceutically acceptable
resistant
bacterial
Prior art date
Application number
PCT/US2012/021270
Other languages
English (en)
Inventor
Arnaud Le Tiran
Anne-Laure Grillot
Paul S. Charifson
Youssef Laafiret BENNANI
Hardwin O'dowd
Emanuele Perola
Original Assignee
Vertex Pharmaceuticals Incorporated
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to AU2012205415A priority Critical patent/AU2012205415B2/en
Priority to SG2013053426A priority patent/SG191946A1/en
Priority to RU2013137750A priority patent/RU2609259C2/ru
Priority to CN201280012742.5A priority patent/CN103443096B/zh
Application filed by Vertex Pharmaceuticals Incorporated filed Critical Vertex Pharmaceuticals Incorporated
Priority to MX2013008162A priority patent/MX339455B/es
Priority to KR1020137018403A priority patent/KR101897952B1/ko
Priority to NZ612961A priority patent/NZ612961B2/en
Priority to CA2824516A priority patent/CA2824516C/fr
Priority to ES12702920.5T priority patent/ES2545516T3/es
Priority to BR112013017974-0A priority patent/BR112013017974B1/pt
Priority to JP2013549575A priority patent/JP6085829B2/ja
Priority to EP12702920.5A priority patent/EP2663557B1/fr
Publication of WO2012097269A1 publication Critical patent/WO2012097269A1/fr
Priority to IL227406A priority patent/IL227406A/en
Priority to ZA2013/05233A priority patent/ZA201305233B/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6558Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system
    • C07F9/65586Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing at least two different or differently substituted hetero rings neither condensed among themselves nor condensed with a common carbocyclic ring or ring system at least one of the hetero rings does not contain nitrogen as ring hetero atom

Definitions

  • Gyrase is one of the topoisomerases, a group of enzymes which catalyze the interconversion of topological isomers of DNA (see generally, Kornberg and Baker, DNA Replication, 2d Ed., Chapter 12, 1992, W. H. Freeman and Co.; Driica, Molecular
  • Gyrase itself controls DNA supercoiling and relieves topological stress that occurs when the DNA strands of a parental duplex are untwisted during the replication process. Gyrase also catalyzes the conversion of relaxed, closed circular duplex DNA to a negatively superhelical form which is more favorable for recombination.
  • the mechanism of the supercoiling reaction involves the wrapping of gyrase around a region of the DNA, double strand breaking in that region, passing a second region of the DNA through the break, and rejoining the broken strands. Such a cleavage mechanism is characteristic of a type II topoisomerase.
  • the supercoiling reaction is driven by the binding of ATP to gyrase.
  • the ATP is then hydrolyzed during the reaction.
  • This ATP binding and subsequent hydrolysis cause conformational changes in the DNA-bound gyrase that are necessary for its activity. It has also been found that the level of DNA supercoiling (or relaxation) is dependent on the ATP/ADP ratio. In the absence of ATP, gyrase is only capable of relaxing supercoiled DNA.
  • Bacterial DNA gyrase is a 400 kilodalton protein tetramer consisting of two A (GyrA) and two B subunits (GyrB). Binding and cleavage of the DNA is associated with GyrA, whereas ATP is bound and hydrolyzed by the GyrB protein. GyrB consists of an amino -terminal domain which has the ATPase activity, and a carboxy-terminal domain which interacts with GyrA and DNA.
  • eukaryotic type II topoisomerases are homodimers that can relax negative and positive supercoils, but cannot introduce negative supercoils.
  • an antibiotic based on the inhibition of bacterial DNA gyrase and/or topoisomerase IV would be selective for these enzymes and be relatively inactive against the eukaryotic type II topoisomerases.
  • Topoisomerase IV primarily resolves linked chromosome dimers at the conclusion of DNA replication.
  • the widely-used quinolone antibiotics inhibit bacterial DNA gyrase (GyrA) and/or Topoisomerase IV (ParC).
  • examples of the quinolones include the early compounds such as nalidixic acid and oxolinic acid, as well as the later, more potent fluoroquinolones such as norfloxacin, ciprofloxacin, and trovafloxacin. These compounds bind to GyrA and/or ParC and stabilize the cleaved complex, thus inhibiting overall gyrase function, leading to cell death.
  • the fluoroquinolones inhibit the catalytic subunits of gyrase (GyrA) and/or Topoisomerase IV (Par C) (see Drlica and Zhao, Microbiology and Molecular Biology Reviews, 1997, 61 , 377-392).
  • GyrA catalytic subunits of gyrase
  • par C Topoisomerase IV
  • drug resistance has also been recognized as a problem for this class of compounds (WHO Report, "Use of Quinolones in Food Animals and Potential Impact on Human Health", 1998).
  • quinolones as with other classes of antibiotics, bacteria exposed to earlier compounds often quickly develop cross-resistance to more potent compounds in the same class.
  • GyrB gyrase
  • ParE topoisomerase IV
  • Compounds that target these same ATP binding sites in the GyrB and ParE subunits would be useful for treating various bacterial infections (see, Charifson et al., J. Med. Chem., 2008, 51, pp. 5243-5263).
  • antibiotics that represent a new class of compounds not previously used to treat bacterial infection.
  • Compounds that target the ATP binding sites in both the GyrB (gyrase) and ParE (topoisomerase IV) subunits would be useful for treating various bacterial infections.
  • Such compounds would be particularly useful in treating nosocomial infections in hospitals where the formation and transmission of resistant bacteria are becoming increasingly prevalent.
  • new antibiotics having a broad spectrum of activity with advantageous toxicological properties.
  • the present invention is directed to compounds and pharmaceutically acceptable salts thereof, useful as gyrase and/or topoisomerase IV inhibitors.
  • the gyrase and/or topoisomerase IV inhibitors of the present invention may be represented by formula (I) or salts thereof:
  • R is hydrogen or fluorine
  • X is hydrogen, -PO(OH) 2 , -PO(OH)0 " M ,
  • the compounds of formula (I) either possess a broad range of anti-bacterial activity and advantageous toxicological properties or are prodrugs of compounds having said activity.
  • the present invention also relates to compounds of formula (IA), or
  • the compounds of formula (IA) are encompassed by formula (I).
  • the compounds of formula (IA) may be represente
  • the present invention also relates to compounds of formula (IB), or
  • the compounds of formula (IB) are encompassed by formula (I).
  • the compounds of formula (IB) may be represented as:
  • the compounds of formula (IB) are phosphate ester prodrugs of the compound (R)-l-ethyl-3-(6-fluoro-5-(2-(2-hydroxypropan-2-yl)pyrimidin-5-yl)-7- (tetrahydrofuran-2-yl)-lH-benzo[d]imidazol-2-yl)urea, which possesses a broad range of antibacterial activity and advantageous toxicological properties.
  • the present invention further provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) (which includes other formulae encompassed by formula (I) such as formulae (IA), (IB), (IC) and (ID)) or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, a pharmaceutically acceptable carrier, and an additional therapeutic agent selected from an antibiotic, an anti-inflammatory agent, a matrix metalloproteinase inhibitor, a lipoxygenase inhibitor, a cytokine antagonist, an immunosuppressant, an anti-cancer agent, an anti-viral agent, a cytokine, a growth factor, an immunomodulator, a prostaglandin or an anti-vascular hyperproliferation compound.
  • the present invention relates to a method of treating a bacterial infection in a mammal in need thereof, comprising administering to said mammal a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • the present invention relates to a method of treating a bacterial infection in a mammal in need thereof, comprising administering to said mammal a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof and an antibiotic, an anti-inflammatory agent, a matrix metalloproteinase inhibitor, a lipoxygenase inhibitor, a cytokine antagonist, an
  • an anti-cancer agent an anti- viral agent, a cytokine, a growth factor, an immunomodulator, a prostaglandin or an anti- vascular hyperproliferation compound, either as part of a multiple dosage form together with said compound or as a separate dosage form.
  • FIG. 1 is a thermal ellipsoid plot of two symmetry independent molecules of compound 12.
  • FIG. 2 is a thermal ellipsoid plot of two symmetry independent molecules of compound 23.
  • halogen means F, CI, Br, or I.
  • structures depicted herein are also meant to include all stereochemical forms of the structure; i.e., the R and S configurations for each asymmetric center. Therefore, single stereochemical isomers as well as enantiomeric and diastereomeric mixtures of the present compounds are within the scope of the invention.
  • Isotopically-labeled forms of compounds of formula (I) wherein one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature are also included herein.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, and fluorine, such as 2 H, 3 H, 13 C, 14 C, 15 N, 18 0, and 17 0.
  • Such radio-labeled and stable-isotopically labeled compounds are useful, for example, as research or diagnostic tools or gyrase and/or topoisomerase IV inhibitors with improved therapeutic profile.
  • the structures also encompass zwitterionic forms of the compounds or salts, where appropriate.
  • compounds of formula (I) include compounds of formula (IC)
  • compounds of formula (I) include compounds of formulae (ID) and (IE) as set forth below:
  • the compounds of formula (IB) are prodrugs of their parent compound, l-ethyl-3- [6-fluoro-5-[2-(l-hydroxy-l-methyl-ethyl)pyrimidin-5-yl]-7-[(2R)-tetrahydrofuran-2-yl]-lH- benzimidazol-2-yl]urea.
  • the activity exhibited upon administration of the prodrug is principally due to the presence of the parent compound that results from cleavage of the prodrug.
  • prodrug refers to compounds which are drug precursors which, following administration and absorption, release the drug in vivo via some metabolic process.
  • a prodrug possesses less biological activity than its parent drug.
  • a prodrug may also improve the physical properties of the parent drug and/or it may also improve overall drug efficacy, for example through the reduction of toxicity and unwanted effects of a drug by controlling its absorption, blood levels, metabolic distribution and cellular uptake.
  • parent compound or “parent drug” refers to the biologically active entity that is released via enzymatic action of a metabolic or a catabolic process, or via a chemical process following administration of the prodrug.
  • the parent compound may also be the starting material for the preparation of its corresponding prodrug.
  • the monovalent cations defined by M + include ammonium, alkali metal ions such as sodium, lithium and potassium ions, dicyclohexylamine ion, and N-methyl-D-glucamine ion.
  • the divalent cations defined by D 2+ include, alkaline earth metal ions such as aluminum, calcium and magnesium ions. Also included are amino acid cations such as ions of arginine, lysine, ornithine, and so forth. If M is a monovalent cation, it is recognized that if the definition 2M + is present, each of M + may be the same or different.
  • the basic nitrogen-containing groups may be quaternized with such agents as: lower alkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl; diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; aralkyl halides like benzyl bromide and others.
  • lower alkyl halides such as methyl, ethyl, propyl, and butyl chloride, bromides and iodides
  • dialkyl sulfates like dimethyl, diethyl, dibutyl
  • diamyl sulfates long chain halides
  • each R 9 is independently hydrogen or a C1-C4 alkyl group
  • the prodrugs of the present invention are characterized by unexpectedly high aqueous solubility. This solubility facilitates administration of higher doses of the prodrug, resulting in a greater drug load per unit dosage.
  • One embodiment of this invention relates to a method of treating a bacterial infection in a mammal in need thereof, comprising administering to said mammal a therapeutically effective amount of a compound having the formula (I) or a pharmaceutically acceptable salt thereof.
  • the invention provides a method of decreasing or inhibiting bacterial quantity in a biological sample.
  • This method comprises contacting said biological sample with a compound of formula (I) or a pharmaceutically acceptable salt thereof.
  • biological sample includes cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
  • biological sample also includes living organisms, in which case "contacting a compound of this invention with a biological sample” is synonymous with the term “administering said compound or composition comprising said compound) to a mammal”.
  • One embodiment comprises contacting said biological sample with a compound selected from the group consisting of (R)-l -ethyl-3-(5-(2-(2-hydroxypropan-2-yl)pyrimidin- 5-yl)-7-(tetrahydrofuran-2-yl)-lH-benzo[d]imidazol-2-yl)urea, or a pharmaceutically acceptable salt thereof; and (R)-l -ethyl-3-(6-fluoro-5-(2-(2-hydroxypropan-2-yl)pyrimidin-5- yl)-7-(tetrahydrofuran-2-yl)-lH-benzo[d]imidazol-2-yl)urea, or a pharmaceutically acceptable salt thereof.
  • Pharmaceutical compositions useful for such methods are described below.
  • One embodiment comprises contacting said biological sample with a phosphate ester prodrug of (R)-l -ethyl-3-(6-fluoro-5-(2-(2-hydroxypropan-2-yl)pyrimidin-5-yl)-7- (tetrahydrofuran-2-yl)-lH-benzo[d]imidazol-2-yl)urea, as defined by formula (IB).
  • compositions useful for such methods are described below.
  • the antimicrobial activity of the compounds of formula (I) may be demonstrated in an antimicrobial susceptibility assay.
  • the details of the conditions used for the antimicrobial susceptibility assays are set forth in the Examples below.
  • compositions for administration to animals or humans may be formulated into pharmaceutical compositions for administration to animals or humans.
  • the term "measurably decrease bacterial quantity”, as used herein means a measurable change in the number of bacteria between a sample containing said inhibitor and a sample containing only bacteria.
  • Another embodiment of this invention relates to a method, as described above, of preventing, controlling, treating or reducing the advancement, severity or effects of a bacterial infection in a mammal in need thereof, but further comprising the step of administering to said mammal an agent which increases the susceptibility of bacterial organisms to antibiotics.
  • the methods of the present invention are useful to treat patients in the veterinarian field including, but not limited to, zoo, laboratory, human companion, and farm animals including primates, rodents, reptiles and birds.
  • animals include, but are not limited to, guinea pigs, hamsters, gerbils, rat, mice, rabbits, dogs, cats, horses, pigs, sheep, cows, goats, deer, rhesus monkeys, monkeys, tamarinds, apes, baboons, gorillas, chimpanzees, orangutans, gibbons, ostriches, chickens, turkeys, ducks, and geese.
  • compositions and methods of this invention will be useful generally for controlling bacterial infections in vivo.
  • bacterial organisms that may be controlled by the compositions and methods of this invention include, but are not limited to the following organisms: Streptococcus pneumoniae, Streptococcus pyogenes, Enterococcus faecalis, Enterococcus faecium, Klebsiella pneumoniae, Enterobacter spp. Proteus spp. Pseudomonas aeruginosa, E. coli, Serratia marcescens, Staphylococcus aureus, Coag. Neg.
  • Staphylococci Haemophilus influenzae, Bacillus anthracis, Mycoplasma pneumoniae, Moraxella catarrhalis, Chlamydophila pneumoniae, Chlamydia trachomatis, Legionella pneumophila, Mycobacterium tuberculosis, Helicobacter pylori, Staphylococcus saprophyticus, Staphylococcus epidermidis, Francisella tularensis, Yersinia pestis, Clostridium difficile, Neisseria gonorrhoeae, Neisseria meningitidis, Mycobacterium avium complex, Mycobacteriumabscessus, Mycobacterium kansasii and Mycobacterium ulcerans.
  • nosocomial and non-nosocomial infections include but are not limited to upper respiratory infections, lower respiratory infections, ear infections, pleuropulmonary and bronchial infections, complicated urinary tract infections, uncomplicated urinary tract infections, intra-abdominal infections, cardiovascular infections, a blood stream infection, sepsis, bacteremia, CNS infections, skin and soft tissue infections, GI infections, bone and joint infections, genital infections, eye infections, or granulomatous infections.
  • bacterial infections include but are not limited to uncomplicated skin and skin structure infections (uSSSI), complicated skin and skin structure infections (cSSSI), catheter infections, pharyngitis, sinusitis, otitis externa, otitis media, bronchitis, empyema, pneumonia, community-acquired bacterial pneumoniae (CABP), hospital-acquired pneumonia (HAP), hospital-acquired bacterial pneumonia, ventilator-associated pneumonia (VAP), diabetic foot infections, vancomycin resistant enterococci infections, cystitis and pyelonephritis, renal calculi, prostatitis, peritonitis, complicated intra-abdominal infections (cIAI) and other inter-abdominal infections, dialysis-associated peritonitis, visceral abscesses, endocarditis, myocarditis, pericarditis, transfusion-associated sepsis, meningitis, encephalitis, brain abscess, osteo
  • non-nosocomial infections is also referred to as community acquired infections.
  • compositions and methods will therefore be useful for controlling, treating or reducing the advancement, severity or effects of community-acquired bacterial pneumoniae (CABP), hospital-acquired pneumonia (HAP), hospital-acquired bacterial pneumonia, ventilator-associated pneumonia (VAP), bacteremia, diabetic foot infections, catheter infections, uncomplicated skin and skin structure infections (uSSSI), complicated skin and skin structure infections (cSSSI), vancomycin resistant enterococci infections or osteomyelitis.
  • CABP community-acquired bacterial pneumoniae
  • HAP hospital-acquired pneumonia
  • VAP ventilator-associated pneumonia
  • bacteremia bacteremia
  • diabetic foot infections catheter infections
  • uSSSI uncomplicated skin and skin structure infections
  • cSSSI complicated skin and skin structure infections
  • vancomycin resistant enterococci infections or osteomyelitis vancomycin resistant enterococci infections or osteomyelitis.
  • compositions and methods will therefore be useful for controlling, treating or reducing the advancement, severity or effects of upper respiratory infections, lower respiratory infections, ear infections, pleuropulmonary and bronchial infections, complicated urinary tract infections, uncomplicated urinary tract infections, intraabdominal infections, cardiovascular infections, a blood stream infection, sepsis, bacteremia, CNS infections, skin and soft tissue infections, GI infections, bone and joint infections, genital infections, eye infections, or granulomatous infections, uncomplicated skin and skin structure infections (uSSSI), complicated skin and skin structure infections (cSSSI), catheter infections, pharyngitis, sinusitis, otitis externa, otitis media, bronchitis, empyema, pneumonia, community-acquired bacterial pneumoniae (CABP), hospital-acquired pneumonia (HAP), hospital-acquired bacterial pneumonia, ventilator-associated pneumonia (VAP), diabetic foot infections, vancomycin resistant enterococci infections
  • the bacterial infection is characterized by the presence of one or more of Streptococcus pneumoniae, Streptococcus pyogenes, Enterococcus faecalis, Enterococcus faecium, Staphylococcus aureus, Coag. Neg. Staphlococci, Bacillus anthracis, Staphylococcus epidermidis, Staphylococcus saprophyticus, or Mycobacterium tuberculosis.
  • the bacterial infection is characterized by the presence of one or more of Streptococcus pneumoniae, Enterococcus faecalis, or Staphylococcus aureus.
  • the bacterial infection is characterized by the presence of one or more of E. coli, Moraxella catarrhalis, or Haemophilus influenzae.
  • the bacterial infection is characterized by the presence of one or more of Clostridium difficile, Neisseria gonorrhoeae, Neisseria meningitidis,
  • Mycobacterium avium complex Mycobacterium abscessus, Mycobacterium kansasii, Mycobacterium ulcerans, Chlamydophila pneumoniae and Chlamydia tracomatis.
  • the bacterial infection is characterized by the presence of one or more of Streptococcus pneumoniae, Staphylococcus epidermidis, Enterococcus faecalis, Staphylococcus aureus,, Clostridium difficile, Moraxella catarrhalis, Neisseria gonorrhoeae, Neisseria meningitidis, Mycobacterium avium complex, Mycobacterium abscessus, Mycobacterium kansasii, Mycobacterium ulcerans, Chlamydophila pneumoniae, Chlamydia trachomatis, Haemophilus influenzae, Streptococcus pyogenes or ⁇ -haemolytic streptococci.
  • the bacterial infection is characterized by the presence ofone or more of Methicillin resistant Staphylococcus aureus, Fluoroquinolone resistant Staphylococcus aureus, Vancomycin intermediate resistant Staphylococcus aureus, Linezolid resistant Staphylococcus aureus, Penicillin resistant Streptococcus pneumoniae, Macrolide resistant Streptococcus pneumoniae, Fluoroquinolone resistant Streptococcus pneumoniae, Vancomycin resistant Enterococcus faecalis, Linezolid resistant Enterococcus faecalis, Fluoroquinolone resistant Enterococcus faecalis, Vancomycin resistant Enterococcus faecium, Linezolid resistant Enterococcus faecium, Fluoroquinolone resistant Enterococcus faecium, Linezolid resistant Enterococcus faecium, Fluoroquinolone resistant Enterococcus fae
  • tuberculosis Rifampin resistant Mycobacterium tuberculosis, Methicillin resistant Coagulase negative staphylococcus, Fluoroquinolone resistant Coagulase negative staphylococcus, Glycopeptide intermediate resistant Staphylococcus aureus, Vancomycin resistant
  • Staphylococcus aureus Hetero vancomycin intermediate resistant Staphylococcus aureus, Hetero vancomycin resistant Staphylococcus aureus, Macrolide-Lincosamide-Streptogramin resistant Staphylococcus, ⁇ -lactam resistant Enterococcus faecalis, ⁇ -lactam resistant Enterococcus faecium, Ketolide resistant Streptococcus pneumoniae, Ketolide resistant Streptococcus pyogenes, Macrolide resistant Streptococcus pyogenes, Vancomycin resistant staphylococcus epidermidis, Fluoroquinolone resistant Neisseria gonorrhoeae, Multidrug Resistant Pseudomonas aeruginosa or Cephalosporin resistant Neisseria gonorrhoeae.
  • the Methicillin resistant Staphylococci are selected from Methicillin resista/?? Staphylococcus aureus, Methicillin resistant
  • Staphylococcus epidermidis or Methicillin resistant Coagulase negative staphylococcus.
  • a form of a compound of formula (I) is used to treat community acquired MRS A (i.e., cMRSA).
  • a form of a compound of formula (I) is used to treat daptomycin resistant organism including, but not limited to, daptomycin resistant
  • the Fluoroquinolone resistant Staphylococci are selected from Fluoroquinolone resistant Staphylococcus aureus, Fluoroquinolone resistant
  • the Glycopeptide resistant Staphylococci are selected from Glycopeptide intermediate resistant Staphylococcus aureus, Vancomycin resistant Staphylococcus aureus, Vancomycin intermediate resistant Staphylococcus aureus, Hetero vancomycin intermediate resistant Staphylococcus aureus, or Hetero vancomycin resistant Staphylococcus aureus.
  • the Macrolide-Lincosamide-Streptogramin resistant Staphylococci is Macrolide-Lincosamide-Streptogramin resistant Staphylococcus aureus.
  • the Linezolid resistant Enterococciare selected from Linezolid resistant Enterococcus faecalis, or Linezolid resistant Enterococcus faecium.
  • the Glycopeptide resistant Enterococci are selected from Vancomycin resistant Enterococcus faecium or Vancomycin resistant
  • the ⁇ -lactam resistant Enterococcus faecalis is ⁇ -lactam resistant Enterococcus faecium.
  • the Penicillin resistant Streptococci is N-(0079] According to another embodiment, the Penicillin resistant Streptococci is N-(0079]
  • the Macrolide resistant Streptococci is Macrolide resistant Streptococcus pneumonia.
  • the Ketolide resistant Streptococci are selected from Macrolide resistant Streptococcus pneumoniae and Ketolide resistant Streptococcus pyogenes.
  • the Fluoroquinolone resistant Streptococci is Fluoroquinolone resistant Streptococcus pneumoniae.
  • the ⁇ -lactam resistant Haemophilus is ⁇ -lactam resistant Haemophilus influenzae.
  • the Fluoroquinolone resistant Haemophilus is Fluoroquinolone resistant Haemophilus influenzae.
  • the Macrolide resistant Haemophilus is Macrolide resistant Haemophilus influenzae.
  • the Macrolide resistant Mycoplasma is Macrolide resistant Mycoplasma pneumoniae.
  • the Isoniazid resistant Mycobacterium is Isoniazid resistant Mycobacterium tuberculosis.
  • the Rifampin resistant Mycobacterium is Rifampin resistant Mycobacterium tuberculosis.
  • the ⁇ -lactam resistant Moraxella is ⁇ -lactam resistant Moraxella catarrhalis.
  • the bacterial infection is characterized by the presence of one or more of the following: Methicillin resistant Staphylococcus aureus, Fluoroquinolone resistant Staphylococcus aureus, Vancomycin intermediate resistant Staphylococcus aureus, Linezolid resistant Staphylococcus aureus, Penicillin resistant Streptococcus pneumoniae, Macrolide resistant Streptococcus pneumoniae, Fluoroquinolone resistant Streptococcus pneumoniae, Vancomycin resistant Enterococcus faecalis, Linezolid resistant Enterococcus faecalis, Fluoroquinolone resistant Enterococcus faecalis,
  • Vancomycin resistant Enterococcus faecium Linezolid resistant Enterococcus faecium, Fluoroquinolone resistant Enterococcus faecium, Ampicillin resistant Enterococcus faecium, Macrolide resistant Haemophilus influenzae, ⁇ -lactam resistant Haemophilus influenzae, Fluoroquinolone resistant Haemophilus influenzae, ⁇ -lactam resistant Moraxella catarrhalis, Methicillin resistant Staphylococcus epidermidis, Methicillin resistant Staphylococcus epidermidis, Vancomycin resistant Staphylococcus epidermidis, Fluoroquinolone resistant Staphylococcus epidermidis, Macrolide resistant Mycoplasma pneumoniae, Isoniazid resistant Mycobacterium tuberculosis, Rifampin resistant Mycobacterium tuberculosis, Fluoroquinolone resistant Neisseria gonorrhoeae or Ce
  • the bacterial infection is characterized by the presence of one or more of the following: Methicillin resistant Staphylococcus aureus, Methicillin resistant Staphylococcus epidermidis, Methicillin resistant Coagulase negative staphylococcus, Fluoroquinolone resistant Staphylococcus aureus, Fluoroquinolone resistant Staphylococcus epidermidis, Fluoroquinolone resistant Coagulase negative staphylococcus, Vancomycin resistant Staphylococcus aureus, Glycopeptide intermediate resistant
  • Staphylococcus aureus Vancomycin resistant Staphylococcus aureus, Vancomycin intermediate resistant Staphylococcus aureus, Hetero vancomycin intermediate resistant Staphylococcus aureus, Hetero vancomycin resistant Staphylococcus aureus, Vancomycin resistant Enterococcus faecium, Vancomycin resistant Enterococcus faecalis, Penicillin resistant Streptococcus pneumoniae, Macrolide resistant Streptococcus pneumoniae, Fluoroquinolone resistant Streptococcus pneumoniae, Macrolide resistant Streptococcus pyogenes, or ⁇ -lactam resistant Haemophilus influenzae.
  • the bacterial infection is characterized by the presence of one or more of the following: Methicillin resistant Staphylococcus aureus, Vancomycin resistant Enterococcus faecium, Vancomycin resistant Enterococcus faecalis, Vancomycin resistant Staphylococcus aureus, Vancomycin intermediate resistant
  • Staphylococcus aureus Hetero vancomycin intermediate resistant Staphylococcus aureus, Hetero vancomycin resistant Staphylococcus aureus, Multidrug Resistant Pseudomonas aeruginosa, Isoniazid resistant Mycobacterium tuberculosis, and Rifampin resistant
  • compositions to treat or prevent the above-identified disorders.
  • a "pharmaceutically acceptable derivative or prodrug” means any one of
  • Particularly favored derivatives or prodrugs are those that increase the bioavailability of the compounds of this invention when such compounds are administered to a mammal (e.g., by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species.
  • Pharmaceutically acceptable prodrugs of the compounds of this invention include, without limitation, esters, amino acid esters, phosphate esters, metal salts and sulfonate esters.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from pharmaceutically acceptable inorganic and organic acids and bases.
  • Suitable acid salts include acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate,
  • Salts derived from appropriate bases include alkali metal (e.g., sodium and potassium), alkaline earth metal (e.g., magnesium), ammonium and N + (Ci-4 alkyl)4 salts.
  • alkali metal e.g., sodium and potassium
  • alkaline earth metal e.g., magnesium
  • ammonium and N + (Ci-4 alkyl)4 salts e.g., sodium and potassium
  • ammonium e.g., sodium and potassium
  • N + (Ci-4 alkyl)4 salts e.g., sodium and potassium
  • compositions of this invention comprise a compound of formula (I) or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
  • Such compositions may optionally comprise an additional therapeutic agent.
  • agents include, but are not limited to, an antibiotic, an anti-inflammatory agent, a matrix
  • metalloprotease inhibitor a lipoxygenase inhibitor, a cytokine antagonist, an
  • an anti-cancer agent an anti- viral agent, a cytokine, a growth factor, an immunomodulator, a prostaglandin or an anti- vascular hyperproliferation compound.
  • pharmaceutically acceptable carrier refers to a non-toxic carrier that may be administered to a patient, together with a compound of this invention, and which does not destroy the pharmacological activity thereof.
  • Pharmaceutically acceptable carriers that may be used in the pharmaceutical compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropylene-block polymers, wool fat and self-emulsifying drug delivery systems (SEDDS) such as alpha-tocopherol, polyethyleneglycol 1000 succinate, or other similar polymeric delivery matrices.
  • SEDDS self-emul
  • pharmaceutically effective amount refers to an amount effective in treating or ameliorating a bacterial infection in a patient.
  • prophylactically effective amount refers to an amount effective in preventing or substantially lessening a bacterial infection in a patient.
  • additional therapeutic agents which are normally administered to treat or prevent that condition, may be administered together with the inhibitors of this invention.
  • therapeutic agents include, but are not limited to, an antibiotic, an anti-inflammatory agent, a matrix metalloprotease inhibitor, a lipoxygenase inhibitor, a cytokine antagonist, an immunosuppressant, an anti-cancer agent, an anti- viral agent, a cytokine, a growth factor, an immunomodulator, a prostaglandin or an anti- vascular hyperproliferation compound.
  • the compounds of this invention may be employed in a conventional manner for controlling bacterial infections levels in vivo and for treating diseases or reducing the advancement or severity of effects which are mediated by bacteria. Such methods of treatment, their dosage levels and requirements may be selected by those of ordinary skill in the art from available methods and techniques.
  • a compound of this invention may be combined with a
  • pharmaceutically acceptable adjuvant for administration to a patient suffering from a bacterial infection or disease in a pharmaceutically acceptable manner and in an amount effective to lessen the severity of that infection or disease.
  • the compounds of this invention may be used in compositions and methods for treating or protecting individuals against bacterial infections or diseases over extended periods of time.
  • the compounds of this invention may be used in compositions and methods for treating or protecting individuals against bacterial infections or diseases over a 1 -2 week period.
  • the compounds of this invention may be used in compositions and methods for treating or protecting individuals against bacterial infections or diseases over a 4-8 week period (for example, in the treatment of patients with or at risk for developing endocarditis or osteomyelitis).
  • the compounds of this invention may be used in compositions and methods for treating or protecting individuals against bacterial infections or diseases over an 8-12 week period.
  • the compounds may be employed in such compositions either alone or together with other compounds of this invention in a manner consistent with the conventional utilization of enzyme inhibitors in pharmaceutical compositions.
  • a compound of this invention may be combined with pharmaceutically acceptable adjuvants conventionally employed in vaccines and administered in prophylactically effective amounts to protect individuals over an extended period of time against bacterial infections or diseases.
  • compounds of formula (I) may be used prophylactically to prevent a bacterial infection.
  • compounds of formula (I) may be used before, during or after a dental or surgical procedure to prevent opportunistic infections such as those encountered in bacterial endocarditis.
  • compounds of formula (I) may be used prophylactically in dental procedures, including but not limited to extractions, periodontal procedures, dental implant placements and endodontic surgery.
  • compounds of formula (I) may be used prophylactically in surgical procedures including but not limited to general surgery, respiratory surgery
  • prevent a bacterial infection means the prophylactic use of an antibiotic, such as a gyrase and/or topoisomerase IV inhibitor of the present invention, to prevent a bacterial infection.
  • Treatment with a gyrase and/or topoisomerase IV inhibitor could be done prophylactically to prevent an infection caused by an organism that is susceptible to the gyrase and/or topoisomerase IV inhibitor.
  • prophylactic treatment could be considered is when an individual is more vulnerable to infection due to, for example, weakened immunity, surgery, trauma, presence of an artificial device in the body (temporary or permanent), an anatomical defect, exposure to high levels of bacteria or possible exposure to a disease-causing pathogen.
  • factors that could lead to weakened immunity include chemotherapy, radiation therapy, diabetes, advanced age, HIV infection, and transplantation.
  • An example of an anatomical defect would be a defect in the heart valve that increases the risk of bacterial endocarditis.
  • artificial devices include artificial joints, surgical pins, catheters, etc.
  • prophylactic use of a gyrase and/or topoisomerase IV inhibitor would be to prevent the spread of a pathogen between individuals (direct or indirect).
  • a specific example of prophylactic use to prevent the spread of a pathogen is the use of a gyrase and/or topoisomerase IV inhibitor by individuals in a healthcare institution (for example a hospital or nursing home).
  • the compounds of formula (I) may also be co-administered with other antibiotics to increase the effect of therapy or prophylaxis against various bacterial infections.
  • the compounds of this invention are administered in combination therapies with other agents, they may be administered sequentially or concurrently to the patient.
  • pharmaceutical or prophylactic compositions according to this invention comprise a combination of a compound of formula (I) and another therapeutic or prophylactic agent.
  • the additional therapeutic agent or agents is an antibiotic selected from a natural penicillin, a penicillinase-resistant penicillin, an antipseudomonal penicillin, an aminopenicillin, a first generation cephalosporin, a second generation cephalosporin, a third generation cephalosporin, a fourth generation cephalosporin, a carbapenem, a cephamycin, a quinolone, a fluoroquinolone, an aminoglycoside, a macrolide, a ketolide, a polymyxin, a tetracycline, a glycopeptide, a streptogramin, an oxazolidinone, a rifamycin, or a sulfonamide.
  • an antibiotic selected from a natural penicillin, a penicillinase-resistant penicillin, an antipseudomonal penicillin, an aminopenicillin, a first generation cephalosporin, a second generation cephalosporin, a third generation ce
  • the additional therapeutic agent or agents is an antibiotic selected from a penicillin, a cephalosporin, a quinolone, an aminoglycoside or an
  • the additional therapeutic agents are selected from a natural penicillin including Benzathine penicillin G, Penicillin G and Penicillin V, from a penicillinase-resistant penicillin including Cloxacillin, Dicloxacillin, Nafcillin and Oxacillin, from a antipseudomonal penicillin including Carbenicillin, Mezlocillin, Pipercillin,
  • Pipercillin/tazobactam Ticaricillin and Ticaricillin/Clavulanate, from an aminopenicillin including Amoxicillin, Ampicillin and Ampicillin/Sulbactam, from a first generation cephalosporin including Cefazolin, Cefadroxil, Cephalexin and Cephadrine, from a second generation cephalosporin including Cefaclor, Cefaclor-CD, Cefamandole, Cefonacid, Cefprozil, Loracarbef and Cefuroxime, from a third generation cephalosporin including Cefdinir, Cefixime, Cefoperazone, Cefotaxime, Cefpodoxime, Ceftazidime, Ceftibuten, Ceftizoxme and Ceftriaxone, from a fourth generation cephalosporin including Cefepime, Ceftaroline and Ceftobiprole, from a Cephamycin including Cefotetan and Cefoxit
  • the additional therapeutic agents are selected from a natural penicillin including Penicillin G, from a penicillinase-resistant penicillin including Nafcillin and Oxacillin, from an antipseudomonal penicillin including Pipercillin/tazobactam, from an aminopenicillin including Amoxicillin, from a first generation cephalosporin including Cephalexin, from a second generation cephalosporin including Cefaclor, Cefaclor-CD and Cefuroxime, from a third generation cephalosporin including Ceftazidime and Ceftriaxone, from a fourth generation cephalosporin including Cefepime, from a carbapenem including Imepenem, Meropenem, Ertapenem, Doripenem, Panipenem and Biapenem,a
  • a solid form of a compound of formula (I) can be administered for the treatment of a gram positive infection.
  • the composition is a solid, liquid (e.g., a suspension), or an iv (e.g., a form of the formula (I) compound is dissolved into a liquid and administered iv) composition.
  • the composition including a formula (I) compound is administered in combination with an additional antibiotic agent, for example, a natural penicillin, a penicillinase-resistant penicillin, an antipseudomonal penicillin, an aminopenicillin, a first generation cephalosporin, a second generation cephalosporin, a third generation
  • an additional antibiotic agent for example, a natural penicillin, a penicillinase-resistant penicillin, an antipseudomonal penicillin, an aminopenicillin, a first generation cephalosporin, a second generation cephalosporin, a third generation
  • cephalosporin a fourth generation cephalosporin, a carbapenem, a cephamycin, a quinolone, a fluoroquinolone, an aminoglycoside, a macrolide, a ketolide, a polymyxin, a tetracycline, a glycopeptide, a streptogramin, an oxazolidinone, a rifamycin, or a sulfonamide.
  • the composition including a solid form of a formula (I) compound is administered orally, and the additional antibiotic agent, for example, a natural penicillin, a penicillinase-resistant penicillin, an antipseudomonal penicillin, an aminopenicillin, a first generation cephalosporin, a second generation cephalosporin, a third generation
  • the additional antibiotic agent for example, a natural penicillin, a penicillinase-resistant penicillin, an antipseudomonal penicillin, an aminopenicillin, a first generation cephalosporin, a second generation cephalosporin, a third generation
  • cephalosporin a fourth generation cephalosporin, a carbapenem, a cephamycin, a quinolone, a fluoroquinolone, an aminoglycoside, a macrolide, a ketolide, a polymyxin, a tetracycline, a glycopeptide, a streptogramin, an oxazolidinone, a rifamycin, or a sulfonamide is administered iv.
  • a solid form of a formula (I) compound can be administered for the treatment of a gram negative infection.
  • the composition is a solid, liquid (e.g., a suspension), or an iv (e.g., a form of a formula (I) compound is dissolved into a liquid and administered iv) composition.
  • the composition including a formula (I) compound is administered in combination with an additional antibiotic agent, selected from a: natural penicillin, a penicillinase-resistant penicillin, an antipseudomonal penicillin, an aminopenicillin, a first generation
  • the composition including a solid form of a formula (I) compound is administered orally, and the additional antibiotic agent, for example, a natural penicillin, a penicillinase-resistant penicillin, an antipseudomonal penicillin, an
  • the additional therapeutic agent is administered iv.
  • the additional therapeutic agents described above may be administered separately, as part of a multiple dosage regimen, from the inhibitor-containing composition. Alternatively, these agents may be part of a single dosage form, mixed together with the inhibitor in a single composition.
  • compositions of this invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • the pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles.
  • the pH of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • the pharmaceutical, compositions may be in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally-acceptable diluent or solvent, for example, as a solution in 1 ,3-butanediol.
  • suitable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as those described in Pharmacopeia Helvetica, or a similar alcohol.
  • compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, and aqueous suspensions and solutions.
  • carriers which are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried corn starch.
  • compositions of this invention may also be administered in the form of suppositories for rectal administration.
  • These compositions can be prepared by mixing a compound of this invention with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • Topical administration of the pharmaceutical compositions of this invention is especially useful when the desired treatment involves areas or organs readily accessible by topical application.
  • the pharmaceutical composition should be formulated with a suitable ointment containing the active components suspended or dissolved in a carrier.
  • Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petroleum, white petroleum, propylene glycol, polyoxyethylene, polyoxypropylene, emulsifying wax and water.
  • the pharmaceutical composition can be formulated with a suitable lotion or cream containing the active compound suspended or dissolved in a carrier.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • the pharmaceutical compositions of this invention may also be topically applied to the lower intestinal tract by rectal suppository formulation or in a suitable enema formulation. Topically-administered transdermal patches are also included in this invention.
  • compositions of this invention may be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance
  • compounds of formula (I) may also be delivered by implantation (e.g., surgically), such as with an implantable or indwelling device.
  • An implantable or indwelling device may be designed to reside either permanently or temporarily in a subject.
  • implantable and indwelling devices include, but are not limited to, contact lenses, central venous catheters and needleless connectors, endotracheal tubes, intrauterine devices, mechanical heart valves, pacemakers, peritoneal dialysis catheters, prosthetic joints, such as hip and knee replacements, tympanostomy tubes, urinary catheters, voice prostheses, stents, delivery pumps, vascular filters and implantable control release compositions.
  • Biofilms can be detrimental to the health of patients with an implantable or indwelling medical device because they introduce an artificial substratum into the body and can cause persistent infections.
  • providing compounds of formula (I) in or on the implantable or indwelling device can prevent or reduce the production of a biofilm.
  • implantable or indwelling devices may be used as a depot or reservoir of compounds of formula (I). Any implantable or indwelling device can be used to deliver compounds of formula (I) provided that a) the device, compounds of formula (I) and any pharmaceutical composition including compounds of formula (I) are biocompatible, and b) that the device can deliver or release an effective amount of compounds of formula (I) to confer a therapeutic effect on the treated patient.
  • the implantable device is a stent.
  • a stent can include interlocked meshed cables.
  • Each cable can include metal wires for structural support and polymeric wires for delivering the therapeutic agent.
  • the polymeric wire can be dosed by immersing the polymer in a solution of the therapeutic agent.
  • the therapeutic agent can be embedded in the polymeric wire during the formation of the wire from polymeric precursor solutions.
  • implantable or indwelling devices can be coated with polymeric coatings that include the therapeutic agent.
  • the polymeric coating can be designed to control the release rate of the therapeutic agent. Controlled release of therapeutic agents can utilize various technologies. Devices are known that have a monolithic layer or coating incorporating a heterogeneous solution and/or dispersion of an active agent in a polymeric substance, where the diffusion of the agent is rate limiting, as the agent diffuses through the polymer to the polymer-fluid interface and is released into the surrounding fluid. In some devices, a soluble substance is also dissolved or dispersed in the polymeric material, such that additional pores or channels are left after the material dissolves. A matrix device is generally diffusion limited as well, but with the channels or other internal geometry of the device also playing a role in releasing the agent to the fluid. The channels can be pre-existing channels or channels left behind by released agent or other soluble substances.
  • Erodible or degradable devices typically have the active agent physically immobilized in the polymer.
  • the active agent can be dissolved and/or dispersed throughout the polymeric material.
  • the polymeric material is often hydrolytically degraded over time through hydrolysis of labile bonds, allowing the polymer to erode into the fluid, releasing the active agent into the fluid.
  • Hydrophilic polymers have a generally faster rate of erosion relative to hydrophobic polymers. Hydrophobic polymers are believed to have almost purely surface diffusion of active agent, having erosion from the surface inwards. Hydrophilic polymers are believed to allow water to penetrate the surface of the polymer, allowing hydrolysis of labile bonds beneath the surface, which can lead to homogeneous or bulk erosion of polymer.
  • the implantable or indwelling device coating can include a blend of polymers each having a different release rate of the therapeutic agent.
  • the coating can include a polylactic acid/polyethylene oxide (PLA-PEO) copolymer and a polylactic acid/polycaprolactone (PLA-PCL) copolymer.
  • the polylactic acid/polyethylene oxide (PLA- PEO) copolymer can exhibit a higher release rate of therapeutic agent relative to the polylactic acid/polycaprolactone (PLA-PCL) copolymer.
  • the relative amounts and dosage rates of therapeutic agent delivered over time can be controlled by controlling the relative amounts of the faster releasing polymers relative to the slower releasing polymers.
  • the proportion of faster releasing polymer can be increased relative to the slower releasing polymer. If most of the dosage is desired to be released over a long time period, most of the polymer can be the slower releasing polymer.
  • the device can be coated by spraying the device with a solution or dispersion of polymer, active agent, and solvent. The solvent can be evaporated, leaving a coating of polymer and active agent. The active agent can be dissolved and/or dispersed in the polymer. In some embodiments, the copolymers can be extruded over the device.
  • Dosage levels of between about 0.01 and about 100 mg/kg body weight per day, preferably between 0.5 and about 75 mg/kg body weight per day and most preferably between about 1 and 50 mg/kg body weight per day of the active ingredient compound are useful in a monotherapy for the prevention and treatment of bacterial infections.
  • the pharmaceutical compositions of this invention will be administered from about 1 to 5 times per day or alternatively, as a continuous infusion.
  • the compositions of the present invention may be administered in a pulsatile formulation.
  • Such administration can be used as a chronic or acute therapy.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • a typical preparation will contain from about 5% to about 95% active compound (w/w).
  • such preparations contain from about 20% to about 80% active compound.
  • compositions of this invention comprise a combination of a compound of formula (I) and one or more additional therapeutic or prophylactic agents
  • both the compound and the additional agent should be present at dosage levels of between about 10% to 80% of the dosage normally administered in a monotherapy regime.
  • a maintenance dose of a compound, composition or combination of this invention may be administered, if necessary.
  • the dosage or frequency of administration, or both may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level, treatment should cease.
  • Patients may, however, require intermittent treatment on a long-term basis upon any recurrence or disease symptoms.
  • the invention provides methods for treating or preventing a bacterial infection, or disease state, comprising the step of administering to a patient any compound, pharmaceutical composition, or combination described herein.
  • patient means an animal, preferably a mammal, and most preferably a human.
  • the compounds of this invention are also useful as commercial reagents which effectively bind to the gyrase B and/or topoisomerase IV enzymes.
  • the compounds of this invention, and their derivatives may be used to block gyrase B and/or topoisomerase IV activity in biochemical or cellular assays for bacterial gyrase B and/or topoisomerase IV or their homologs or may be derivatized to bind to a stable resin as a tethered substrate for affinity chromatography applications.
  • the compounds of this invention may be prepared in accordance with general methods known to those skilled in the art for analogous compounds, as taught by U.S. Pat. No. RE40245 E; U.S. Pat. No. 7,495,014 B2; U.S. Pat. No. 7,569,591 B2; U. S. Pat. No. 7,582,641 B2; U.S. Pat. No. 7,618,974 B2; and U.S. Pat. No. 7,727,992 B2. All six of said patents are incorporated by reference as if fully set forth herein. The details of the conditions used for preparing the compounds of the present invention are further set forth in the Examples.
  • NADH nicotinamide adenine dinucleotide (reduced form)
  • VMAX the maximum initial velocity or rate of a reaction
  • reaction temperature rose to 40°C over about 30 minutes at which time a cold water bath was used to control the exotherm and bring the reaction to room temperature. The cold bath was then removed and stirring continued for another 40 minutes (HPLC showed very little remaining un-nitrated material).
  • the reaction mixture was slowly poured into a stirring mixture of crushed ice (800 g). The solid precipitate was collected by filtration, washed with water, saturated aqueous NaHCOs (to pH 8), water again, and hexane. The wet solid was dried first in a convection oven at 50°C for several hours and then under reduced pressure in an oven at 40°C overnight giving (6) as a light brown solid (77.86 g, 62% yield; 98% HPLC purity).
  • Example 7 [00160] Alternate preparation of 2-[5-(4-amino-3-nitro-5-tetrahydrofuran-2-yl- phenyl)pyrimidin-2-yl]propan-2-ol (8).
  • a diffraction data set of reciprocal space was obtained to a resolution of 0.9 A using 0.5° steps using 60 s exposure for each frame. Data were collected at 100 (2) K.
  • Analytical chiral HPLC shows one enantiomer with retention time of 6.3 min eluting with DCM / MeOH / TEA (60 / 40 / 0.1) on a CHIRALPAK® IC® 4.6 x 250 mm column with 1 mL/min flow rate at 30°C.
  • LCMS (CI 8 column eluting with 10-90% CH 3 CN / water gradient over 5 minutes with formic acid modifier) M+l : 411.53 (1.74 min).
  • the minor product was 2-(2- fluoro-6-nitro-phenyl)-2,5-dihydrofuran (15B) (4%): GCMS (Agilent HP-5 MS 30 m x 250 ⁇ x 0.25 ⁇ column heating at 60°C for 2 min to 300°C over 15 min with a 1 mL/min flow rate) M+1 : 210 (11.95 min).
  • the desired filtrate was concentrated on a rotary evaporator to a thick suspension, and the solid was collected by filtration, triturated with MTBE, and dried in vacuo giving 20 as a bright yellow solid (55.8% yield, 90-97% HPLC purity).
  • the filtrate was concentrated and the above purification was repeated giving a second crop of 20 as a bright yellow solid (19.7% yield).
  • the filtrate was again concentrated giving a dark brown oil and this was loaded on a silica column with toluene and minimal CH2CI2. It was eluted with EtOAc / hexane (0% to 50%).
  • the desired fractions were concentrated to a slurry and diluted with MTBE / hexane.
  • Analytical chiral HPLC retention time was 7.7 min (CHIRALPAK® IC® 4.6 x 250 mm column, 1 mL/min flow rate, 30°C). The solid was suspended in 2: 1 EtOH / Et 2 0 (5 volumes), stirred for 10 minutes, collected by filtration, washed with 2: 1 EtOH / Et 2 0, and dried under reduced pressure giving a white solid (60.6 g).
  • a diffraction data set of reciprocal space was obtained to a resolution of 0.85 A using 0.5° steps using 30 s exposures for each frame. Data were collected at 100 (2) K. Integration of intensities and refinement of cell parameters were accomplished using APEXII software. Observation of the crystal after data collection showed no signs of decomposition. As shown in Fig. 2, there are two symmetry independent molecules in the structure and both symmetry independent molecules are R isomers.
  • Analytical chiral HPLC shows one enantiomer with retention time of 8.6 min eluting with CH 2 C1 2 / MeOH / TEA (60 / 40 / 0.1) on a CHIRALPAK® IC® 4.6 x 250 mm column with 1 mL/min flow rate at 30°C.
  • a second crop of white solid product 23A (4.36 g, 98% HPLC purity, 99+% ee) was obtained from the filtrate.
  • LCMS (CI 8 column eluting with 10-90% CH 3 CN / water gradient over 5 minutes with formic acid modifier) M+l : 429.58 (2.03 min).
  • the (R)-l-ethyl-3-(6-fluoro-5-(2-(2-hydroxypropan-2-yl)pyrimidin-5-yl)-7- (tetrahydrofuran-2-yl)-lH-benzo[d]imidazol-2-yl)urea 23 may then be converted to the phosphate or phosphate salt prodrugs according to the schemes set forth below.
  • Reagents and conditions (a) dibenzyl N,N-diisopropylphosphoramidite, tetrazole, 23 °C, DMF; (b) mCPBA, 0-23 °C, DMF; (c) H 2 , Pd/C, M + OH " or D 2+ (OH " ) 2 , EtOH, H 2 0; (d) aq H + ; (e) aq M + OH " .
  • Compounds of formula (IB) may be prepared from compound 23 as shown in Scheme 1.
  • Step 1 compound 23 is treated with dibenzyl N,N-diisopropylphosphoramidite and tetrazole, followed by meta-chloroperoxybenzoic acid (mCPBA), to afford dibenzyl phosphate 24.
  • Reagents and conditions (a) Boc 2 0, DMF, 23 °C;(b)dibenzyl N,N- diisopropylphosphoramidite, tetrazole, 23 °C, DMF; (c) mCPBA, 0-23 °C, DMF; (d) TFA, H 2 0, MeOH, DCM, 23 °C; (e) H 2 , Pd/C, M + OH " or D 2+ (OH ) 2 , EtOH, H 2 0; (f) aq H + ; (g) aq M OH " .
  • the compounds of formula (IB) may be prepared from compound 23 as shown in Scheme 2.
  • Step 1 compound 23 is treated with di-tert-butyl di carbonate (Boc 2 0) to afford protected benzimidazole compound 25.
  • Step 2 compound 25 is treated with dibenzyl N,N-diisopropylphosphoramidite and tetrazole, followed by mCPBA, to afford protected dibenzyl phosphate 26.
  • compound 26 is treated with trifluoro acetic acid (TFA) to remove the protecting group and afford dibenzyl phosphate 24.
  • TFA trifluoro acetic acid
  • the compounds of formula (IB) may also be prepared from compound 23 as shown in Scheme 3.
  • Step 1 compound 23 is treated with two equivalents of Boc 2 0 in the presence of ⁇ , ⁇ -dimethylaminopyridine (DMAP) to afford bis-protected benzimidazole compound 28.
  • Step 2 compound 28 is treated with dibenzyl N,N- diisopropylphosphoramidite and tetrazole, followed by mCPBA, to afford bis-protected dibenzyl phosphate 29.
  • compound 29 is treated with TFA to remove the protecting groups.
  • the resulting filtrate was passed through a pad of Celite and the dark material did not elute until the pad was rinsed with water.
  • the resulting dark solution (approx. 700 mL) was diluted with three volumes of EtOH (2100 mL), filtered through a 0.22 ⁇ PES membrane (using 4 disposable Corning polystyrene filter systems, #431098) and the filtrate concentrated in vacuo.
  • the resulting residue was dissolved in water (100 mL) and EtOH (300 mL), filtered through a 0.22 ⁇ PES membrane to give a clear yellow solution, then passed through a Celite plug (26 mm diameter x 60 mm height, pre-wet with EtOH) rinsing with EtOH (50 mL) and the filtrate then concentrated.
  • the resulting residue was dissolved in water (250 mL) in a 1 L round bottom flask, then 1 M aqueous HC1 (40 mL) was slowly added over 15 min with stirring to give a slurry of white solid. Twenty minutes following completion of the HC1 addition, the solid was collected via filtration through a 0.22 ⁇ PES membrane.
  • Example 26 [00232] Preparation of (i?)-dibenzyl 2-(5-(2-(3-ethylureido)-6-fluoro-7- (tetrahydrofuran-2-yl)-lH-benzo[ii]imidazol-5-yl)pyrimidin-2-yl)propan-2-yl phosphate (24).
  • the suspension was evacuated for 3 min (needle to pump) then placed under an atmosphere of hydrogen gas (balloon). After stirring 2.5 h at 23 °C, the reaction was filtered through a 0.22 ⁇ PES membrane (disposable Corning filter system, 1 L, polystyrene, #431098) to remove catalyst and washed with EtOH (50 mL).
  • reaction mixture was diluted with water and EtOAc (300 mL each), the organic layer separated, washed with water and brine (300 mL each), then dried over magnesium sulfate, filtered and concentrated.
  • the residue was purified by MPLC using an ISCO COMBIFLASH brand flash chromatography purification system (80 g silica column) eluting with a 0-60% EtOAc in hexanes linear gradient over 20 column volumes at 60 mL/min flow rate.
  • the residue was purified by MPLC using an ISCO COMBIFLASH brand flash chromatography purification system (80 g silica column) eluting with a 0-80% EtOAc in hexanes linear gradient over 20 column volumes at 60 mL/min flow rate.
  • Example 30 [00244] Preparation of sodium (i?)-2-(5-(2-(3-ethylureido)-6-fluoro-7- (tetrahydrofuran-2-yl)-lH-benzo[d]imidazol-5-yl)pyrimidin-2-yl)propan-2-yl phosphate (W).
  • the resulting white solid was re-subjected to a MeOH slurry (3 mL), then collected via filtration to give 68 mg of white solid after drying.
  • the white solid was treated with 0.10 M aq NaOH (2.68 mL, 2 equiv NaOH) to give a solution that was then passed through an Acrodisc CR 13 mm syringe filter with 0.45 ⁇ PTFE membrane, flushing with water (2 mL).
  • Protocol #1 Gyrase MIC determination of compounds using microdilution broth method
  • E. faecalis E. faecium and S. pneumoniae: 14.1 mL MHII + 0.9 mL laked horse blood was used.
  • Protocol 2 Gyrase MIC determination of compounds against Gram negatives using microdilution broth method
  • Protocol #3 Gyrase MIC determination of compounds using Agar method
  • Clostridium difficile ATCC BAA- 1382 1. Clostridium difficile ATCC BAA- 1382 ;
  • Routine test concentrations were: 0.002 ug/mL - 16 ug/mL (14 plates).
  • VWR 90003-636 required 2 anaerobic sachets (VWR 90003- 642), while the tall cylinder style chambers (VWR 90003-602) only required 1 sachet.
  • inoculated last after the MIC test plates.
  • the former and latter served as growth and inoculation controls.
  • Table 1 Compound dilutions for MIC determination using the agar dilution method.
  • 1,600 ug/ml 64 ul (lOmg/ml stock) + 336 ul DMSO; 400 ul total volume to start *compound dissolved and diluted in 100%
  • Protocol 4 MIC Determination Procedure for Mycobacterium species
  • Inoculum Preparation for M. tuberculosis 1. Used prepared frozen M. tuberculosis stock stored at -70 C. M. tuberculosis was grown in 7H10 broth + 10% OADC, then frozen at a concentration of 100 Klett or 5 x 10 7 cfu/ml,
  • Protocol 5 Protocol for Mycobacterium tuberculosisSerum Shift MIC Assay
  • Control drug stocks Isoniazid and Novobiocin were prepared at 10 mM in 100% DMSO while Ciprofloxacin and Rifampin were prepared at 1 mM in 50% DMSO and 100% DMSO, respectively.
  • concentrations decreased in 2x steps moving across the rows of the microtiter plate. All MICs were done in duplicate at each of the three medium conditions.
  • Test sets of compounds were typically at 10 mM and 50 ⁇ volume.
  • control compounds generated 2-fold, 11 -point dilutions of each compound using DMSO as diluent. In all cases, left column 12 as DMSO only for a control. Once all dilutions were complete, again transferred 1 ⁇ of each dilution into an empty microtiter well prior to the addition of 100 ⁇ of cells as done for the control compounds. 6. All wells were inoculated with 100 ⁇ of diluted cell suspension (see above).
  • Table 2 shows the results of the MIC assay for selected compounds of this invention.
  • Compound 23 corresponds to l -ethyl-3-[6-fluoro-5-[2-(l - hydroxy-l -methyl-ethyl)pyrimidin-5-yl]-7-[(2R)-tetrahydrofuran-2-yl]-lH-benzimidazol-2- yl]urea and "Compound 23 A” relates to the mesylate salt of Compound 23. These are the same numbers used to identify said compounds and salts as used in the Examples above.
  • Staphylococcus aureus 1 0.13 0.021
  • Staphylococcus aureus 1 2 0.5
  • Staphylococcus aureus 1 1.29 0.3
  • Haemophilus influenzae 2 1 0.5 ATCC 49247
  • Haemophilus influenzae (Rdl 2 2.5 1.3 KW20) ATCC 51907
  • Neisseria gonorrhoeae 3 1.3 0.42 ATCC 35541
  • Escherichia coli CHE30 2 >16 8 Escherichia coli CHE30 tolC 2 0.5 0.125 Escherichia coli MC4100 2 >16 >16 Escherichia coli MC4100 2 1 0.25 tolC
  • Table 3 shows the results of the MIC90 assay for selected compounds of this invention.
  • Neisseria 13 3 0.5- 1 1 0.12- 0.25 gonorrhoeae 0.25
  • Table 3A also shows the results of the MIC assays for selected compounds of this invention.
  • “Compound 23 A” corresponds to the methanesulfonic acid salt of l-ethyl-3-[6-fluoro-5-[2-(l-hydroxy-l-methyl-ethyl)pyrimidin-5-yl]-7-[(2i?)- tetrahydrofuran-2-yl]-lH-benzimidazol-2-yl]urea (23A).
  • Compound W corresponds to disodium (i?)-2-(5-(2-(3-ethylureido)-6-fluoro-7-(tetrahydrofuran-2-yl)-lH- benzo[d]imidazol-5-yl)pyrimidin-2-yl)propan-2-yl phosphate (W). These are the same numbers used to identify said compounds in the synthetic Examples above.
  • CMI Clinical Microbiology Institute located in Wilsonville, Oregon.

Abstract

L'invention concerne un composé de formule (I) : ou un sel de celui-ci pharmaceutiquement acceptable ; X et R sont tels que définis dans le descriptif. Les composés de formule (I) sont utilisés comme inhibiteurs de gyrase et/ou topoïsomérase IV pour traiter des infections bactériennes. Les composés de formule (I) possèdent un large spectre d'activité antibactérienne et des propriétés toxicologiques avantageuses ; ils sont également des promédicaments de composés ayant ladite activité.
PCT/US2012/021270 2011-01-14 2012-01-13 Inhibiteurs de pyrimidine gyrase et topoïsomérase iv WO2012097269A1 (fr)

Priority Applications (14)

Application Number Priority Date Filing Date Title
KR1020137018403A KR101897952B1 (ko) 2011-01-14 2012-01-13 피리미딘 자이라제 및 토포이소머라제 iv 억제제
RU2013137750A RU2609259C2 (ru) 2011-01-14 2012-01-13 Пиримидиновые ингибиторы гиразы и топоизомеразы iv
CN201280012742.5A CN103443096B (zh) 2011-01-14 2012-01-13 促旋酶和拓扑异构酶iv抑制剂
CA2824516A CA2824516C (fr) 2011-01-14 2012-01-13 Inhibiteurs de pyrimidine gyrase et topoisomerase iv
MX2013008162A MX339455B (es) 2011-01-14 2012-01-13 Inhibidores de pirimidina y topoisomerasa iv.
SG2013053426A SG191946A1 (en) 2011-01-14 2012-01-13 Pyrimidine gyrase and topoisomerase iv inhibitors
NZ612961A NZ612961B2 (en) 2011-01-14 2012-01-13 Pyrimidine gyrase and topoisomerase iv inhibitors
AU2012205415A AU2012205415B2 (en) 2011-01-14 2012-01-13 Pyrimidine gyrase and topoisomerase IV inhibitors
ES12702920.5T ES2545516T3 (es) 2011-01-14 2012-01-13 Inhibidores de la pirimidina girasa y la topoisomerasa IV
BR112013017974-0A BR112013017974B1 (pt) 2011-01-14 2012-01-13 inibidores de pirimidina girase e topoisomerase iv
JP2013549575A JP6085829B2 (ja) 2011-01-14 2012-01-13 ピリミジンジャイレースおよびトポイソメラーゼiv阻害剤
EP12702920.5A EP2663557B1 (fr) 2011-01-14 2012-01-13 Inhibiteurs de pyrimidine gyrase et topoïsomérase iv
IL227406A IL227406A (en) 2011-01-14 2013-07-10 Pyrimidine Gyrase and Topoisemarase Suppressors IV preparations containing them and their uses
ZA2013/05233A ZA201305233B (en) 2011-01-14 2013-07-11 Pyrimidine gyrase and topoisomerase iv inhibitors

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201161432965P 2011-01-14 2011-01-14
US61/432,965 2011-01-14
US201161499134P 2011-06-20 2011-06-20
US61/499,134 2011-06-20
US201161515174P 2011-08-04 2011-08-04
US201161515249P 2011-08-04 2011-08-04
US61/515,174 2011-08-04
US61/515,249 2011-08-04

Publications (1)

Publication Number Publication Date
WO2012097269A1 true WO2012097269A1 (fr) 2012-07-19

Family

ID=45567120

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/021270 WO2012097269A1 (fr) 2011-01-14 2012-01-13 Inhibiteurs de pyrimidine gyrase et topoïsomérase iv

Country Status (18)

Country Link
US (2) US8481551B2 (fr)
EP (1) EP2663557B1 (fr)
JP (1) JP6085829B2 (fr)
KR (1) KR101897952B1 (fr)
CN (1) CN103443096B (fr)
AR (1) AR084863A1 (fr)
AU (1) AU2012205415B2 (fr)
BR (1) BR112013017974B1 (fr)
CA (1) CA2824516C (fr)
CL (1) CL2013002025A1 (fr)
ES (1) ES2545516T3 (fr)
IL (1) IL227406A (fr)
MX (1) MX339455B (fr)
RU (1) RU2609259C2 (fr)
SG (1) SG191946A1 (fr)
TW (1) TWI546298B (fr)
WO (1) WO2012097269A1 (fr)
ZA (1) ZA201305233B (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012177707A1 (fr) * 2011-06-20 2012-12-27 Vertex Pharmaceuticals Incorporated Esters phosphoriques d'inhibiteurs de gyrases et de topoisomérases
WO2014014845A1 (fr) * 2012-07-18 2014-01-23 Vertex Pharmaceuticals Incorporated Thérapie combinée comprenant de la 1-éthyl-3[5-[-2-{1-hydroxy-1-méthyl-éthyl}pyrimidin-5-yl]-7-(tétrahydrofuran-2-yl}-1h-benzimidazol-2-yl]urée et ses dérivés pour traiter des maladies mycobactériennes
WO2014015105A1 (fr) * 2012-07-18 2014-01-23 Vertex Pharmaceuticals Incorporated Formes solides de (r)-2-(5-(2-(3-éthylureido)-6-fluoro-7-(tétrahydrofuran-2-yl)-1h-benzo[d]imidazol-5-yl)pyrimidin-2-yl)propan-2-yl dihydrogen phosphate et ses sels
US8912326B2 (en) 2011-01-14 2014-12-16 Vertex Pharmaceuticals Incorporated Process of making gyrase and topoisomerase inhibitors
US8969359B2 (en) 2011-01-14 2015-03-03 Vertex Pharmaceuticals Incorporated Gyrase and topoisomerase IV inhibitors
US9040542B2 (en) 2011-01-14 2015-05-26 Vertex Pharmaceuticals Incorporated Solid forms of gyrase inhibitor (R)-1-ethyl-3-[6-fluoro-5-[2-(1-hydroxy-1-methyl-ethyl)pryimidin-5-yl]-7-(tetrahydrofuran-2-yl)-1H-benzimidazol-2-yl]urea
WO2015114452A2 (fr) 2014-02-03 2015-08-06 Biota Europe Ltd Composés antibactériens
WO2018174288A1 (fr) 2017-03-24 2018-09-27 大正製薬株式会社 Dérivé de 2(1h)-quinolinone

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX351555B (es) * 2011-01-14 2017-10-19 Spero Trinem Inc Formas solidas de inhibidor de girasa (r)-1-etil-3-[5-[2-(1-hidrox i-1-metil-etil)pirimidin-5-il]-7-(tetrahidrofuran-2-il)-1h-benzim idazol-2-il]urea.
BR112019000290B1 (pt) * 2016-07-07 2022-10-18 Cyclerion Therapeutics, Inc Prófármacos de fósforo dos estimuladores da sgc
US20200345637A1 (en) * 2018-01-19 2020-11-05 Aiviva Biopharma, Inc. Suspension compositions of multi-target inhibitors
CN109464472B (zh) * 2019-01-17 2023-09-15 广西医科大学 一种铜绿假单胞菌家兔脓胸引流管生物膜模型建立方法
CN109464471B (zh) * 2019-01-17 2023-09-19 广西医科大学 一种铜绿假单胞菌家兔脓胸脓絮状物生物膜模型建立方法
WO2023076369A1 (fr) * 2021-10-26 2023-05-04 Spero Therapeutics, Inc. Dose efficace humaine et programme de dosage de spr720

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5523288A (en) 1993-09-22 1996-06-04 Xoma Corporation Method of treating gram-negative bacterial infection by administration of bactericidal/permeability-increasing (BPI) protein product and antibiotic
US5783561A (en) 1994-01-14 1998-07-21 Xoma Corporation Anti-gram-positive bacterial methods and materials
US6322847B1 (en) 1999-05-03 2001-11-27 Boston Scientific, Inc. Medical device coating methods and devices
US6569195B2 (en) 1999-07-02 2003-05-27 Scimed Life Systems, Inc. Stent coating
US20030204168A1 (en) 2002-04-30 2003-10-30 Gjalt Bosma Coated vascular devices
US20030216699A1 (en) 2000-05-12 2003-11-20 Robert Falotico Coated medical devices for the prevention and treatment of vascular disease
US20030225450A1 (en) 2001-11-05 2003-12-04 Shulze John E. Drug-delivery endovascular stent and method for treating restenosis
US20030229390A1 (en) 2001-09-17 2003-12-11 Control Delivery Systems, Inc. On-stent delivery of pyrimidines and purine analogs
US20040018228A1 (en) 2000-11-06 2004-01-29 Afmedica, Inc. Compositions and methods for reducing scar tissue formation
US20040044405A1 (en) 2001-10-25 2004-03-04 Wolff Matthew R. Vascular stent or graft coated or impregnated with protein tyrosine kinase inhibitors and method of using same
WO2005012292A1 (fr) * 2003-01-31 2005-02-10 Vertex Pharmaceuticals Incorporated Inhibiteurs de gyrase et utilisations de ces inhibiteurs
WO2006022773A1 (fr) * 2004-07-29 2006-03-02 Vertex Pharmaceuticals Incorporated Inhibiteurs de gyrase et utilisations de ceux-ci
USRE40245E1 (en) 2000-12-15 2008-04-15 Vertex Pharmaceuticals Incorporated Gyrase inhibitors and uses thereof
US7582641B2 (en) 2003-01-31 2009-09-01 Vertex Pharmaceuticals Incorporated Gyrase inhibitors and uses thereof
US7618974B2 (en) 2003-01-31 2009-11-17 Vertex Pharmaceuticals Incorporated Gyrase inhibitors and uses thereof
US7727992B2 (en) 2002-06-13 2010-06-01 Vertex Pharmaceuticals Incorporated Gyrase inhibitors and uses thereof

Family Cites Families (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2015676A1 (de) 1970-04-02 1971-10-21 Farbenfabriken Bayer Ag, 5090 Leverkusen Neue Imine der 2-Formyl-chinoxalindi-N-oxidcarbon-säure-(3) und deren Salze, Verfahren zu ihrer Herstellung und ihre Verwendung als antimikrobielle Mittel
US4174400A (en) 1978-09-13 1979-11-13 Merck & Co., Inc. Anthelmintic benzimidazoles
US4512998A (en) 1980-10-20 1985-04-23 Schering Corporation Anthelmintic benzimidazole carbamates
CA2028530A1 (fr) 1989-11-21 1991-05-22 Christian Hubschwerlen Derives substitues du pyrimidobenzimidazole
DE19514313A1 (de) 1994-08-03 1996-02-08 Bayer Ag Benzoxazolyl- und Benzothiazolyloxazolidinone
HRP960159A2 (en) 1995-04-21 1997-08-31 Bayer Ag Benzocyclopentane oxazolidinones containing heteroatoms
US5643935A (en) 1995-06-07 1997-07-01 The University Of North Carolina At Chapel Hill Method of combatting infectious diseases using dicationic bis-benzimidazoles
TW538046B (en) 1998-01-08 2003-06-21 Hoechst Marion Roussel Inc Aromatic amides having antiobiotic activities and the preparation processes, intermediates and pharmaceutical composition thereof
AUPP873799A0 (en) 1999-02-17 1999-03-11 Fujisawa Pharmaceutical Co., Ltd. Pyridine compounds
GB9911594D0 (en) 1999-05-19 1999-07-21 Smithkline Beecham Plc Novel compounds
GB9912413D0 (en) 1999-05-28 1999-07-28 Pfizer Ltd Compounds useful in therapy
CN101538263A (zh) * 2002-06-13 2009-09-23 沃泰克斯药物股份有限公司 作为促旋酶和/或拓扑异构酶iv的抑制剂用于治疗细菌感染的2-脲基-6-杂芳基-3h-苯并咪唑-4-羧酸衍生物和相关化合物
US8193352B2 (en) 2003-01-31 2012-06-05 Vertex Pharmaceuticals Incorporated Gyrase inhibitors and uses thereof
US8404852B2 (en) 2003-01-31 2013-03-26 Vertex Pharmaceuticals Incorporated Gyrase inhibitors and uses thereof
WO2006092599A2 (fr) * 2005-03-04 2006-09-08 Astrazeneca Ab Composes chimiques
CA2626685A1 (fr) * 2005-11-07 2007-05-18 Vertex Pharmaceuticals Incorporated Derives de benzimidazole utilises en tant qu'inhibiteurs de la gyrase
GB0724349D0 (en) 2007-12-13 2008-01-30 Prolysis Ltd Antibacterial agents
GB0612428D0 (en) 2006-06-22 2006-08-02 Prolysis Ltd Antibacterial agents
US8481544B2 (en) 2006-06-22 2013-07-09 Biota Europe Limited Antibacterial compositions
CN101627038A (zh) 2006-12-04 2010-01-13 阿斯利康(瑞典)有限公司 抗菌多环脲化合物
GB0724342D0 (en) 2007-12-13 2008-01-30 Prolysis Ltd Anitbacterial compositions
US20110166088A1 (en) 2008-06-25 2011-07-07 Ranbaxy Laboratories Limited Benzothiazoles and aza-analogues thereof use as antibacterial agents
EP2475666A2 (fr) 2009-09-11 2012-07-18 Trius Therapeutics, Inc. Inhibiteurs de gyrase
MY176900A (en) 2009-10-16 2020-08-26 Rib X Pharmaceuticals Inc Antimicrobial compounds and methods of making and using the same
CN103249732B (zh) 2010-10-08 2016-08-10 生物区欧洲有限公司 抑制2-乙基氨甲酰氨基-1,3-苯并噻唑-5-基的细菌拓扑异构酶ii
CA2824519C (fr) 2011-01-14 2020-06-16 Vertex Pharmaceuticals Incorporated Procede de fabrication d'inhibiteurs de gyrase et topoisomerase
MX351555B (es) * 2011-01-14 2017-10-19 Spero Trinem Inc Formas solidas de inhibidor de girasa (r)-1-etil-3-[5-[2-(1-hidrox i-1-metil-etil)pirimidin-5-il]-7-(tetrahidrofuran-2-il)-1h-benzim idazol-2-il]urea.
CA2824516C (fr) 2011-01-14 2019-02-26 Vertex Pharmaceuticals Incorporated Inhibiteurs de pyrimidine gyrase et topoisomerase iv
MX341342B (es) * 2011-01-14 2016-08-17 Vertex Pharma Formas solidas del inhibidor de girasa (r)-1-etil-3-[6-fluoro-5-[2 -(1-hidroxi-1-metil-etil) pirimidin-5-il]-7-(tetrahidrofuran-2-il) -1h-benzimidazol-2-il]urea.
TWI554515B (zh) * 2011-06-20 2016-10-21 維泰克斯製藥公司 旋轉酶(gyrase)及拓樸異構酶抑制劑之磷酸酯
WO2013138860A1 (fr) 2012-03-22 2013-09-26 Biota Europe Limited Composés antibactériens
US9018216B2 (en) * 2012-07-18 2015-04-28 Vertex Pharmaceuticals Incorporated Solid forms of (R)-2-(5-(2-(3-ethylureido)-6-fluoro-7-(tetrahydrofuran-2-yl)-1H-benzo[d]imidazol-5-yl)pyrimidin-2-yl)propan-2-yl dihydrogen phosphate and salts thereof
US9572809B2 (en) * 2012-07-18 2017-02-21 Spero Trinem, Inc. Combination therapy to treat Mycobacterium diseases

Patent Citations (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5523288A (en) 1993-09-22 1996-06-04 Xoma Corporation Method of treating gram-negative bacterial infection by administration of bactericidal/permeability-increasing (BPI) protein product and antibiotic
US6140306A (en) 1993-09-22 2000-10-31 Xoma Corporation Method of treating gram-negative bacterial infection by administration of bactericidal/permeability-increasing (BPI) protein product and antibiotic
US5783561A (en) 1994-01-14 1998-07-21 Xoma Corporation Anti-gram-positive bacterial methods and materials
US6322847B1 (en) 1999-05-03 2001-11-27 Boston Scientific, Inc. Medical device coating methods and devices
US6569195B2 (en) 1999-07-02 2003-05-27 Scimed Life Systems, Inc. Stent coating
US20030216699A1 (en) 2000-05-12 2003-11-20 Robert Falotico Coated medical devices for the prevention and treatment of vascular disease
US20040018228A1 (en) 2000-11-06 2004-01-29 Afmedica, Inc. Compositions and methods for reducing scar tissue formation
USRE40245E1 (en) 2000-12-15 2008-04-15 Vertex Pharmaceuticals Incorporated Gyrase inhibitors and uses thereof
US20030229390A1 (en) 2001-09-17 2003-12-11 Control Delivery Systems, Inc. On-stent delivery of pyrimidines and purine analogs
US20040044405A1 (en) 2001-10-25 2004-03-04 Wolff Matthew R. Vascular stent or graft coated or impregnated with protein tyrosine kinase inhibitors and method of using same
US20030225450A1 (en) 2001-11-05 2003-12-04 Shulze John E. Drug-delivery endovascular stent and method for treating restenosis
US20030204168A1 (en) 2002-04-30 2003-10-30 Gjalt Bosma Coated vascular devices
US7727992B2 (en) 2002-06-13 2010-06-01 Vertex Pharmaceuticals Incorporated Gyrase inhibitors and uses thereof
WO2005012292A1 (fr) * 2003-01-31 2005-02-10 Vertex Pharmaceuticals Incorporated Inhibiteurs de gyrase et utilisations de ces inhibiteurs
US7495014B2 (en) 2003-01-31 2009-02-24 Vertex Pharmaceuticals Incorporated Gyrase inhibitors and uses thereof
US7569591B2 (en) 2003-01-31 2009-08-04 Vertex Pharmaceuticals Incorporated Gyrase inhibitors and uses thereof
US7582641B2 (en) 2003-01-31 2009-09-01 Vertex Pharmaceuticals Incorporated Gyrase inhibitors and uses thereof
US7618974B2 (en) 2003-01-31 2009-11-17 Vertex Pharmaceuticals Incorporated Gyrase inhibitors and uses thereof
WO2006022773A1 (fr) * 2004-07-29 2006-03-02 Vertex Pharmaceuticals Incorporated Inhibiteurs de gyrase et utilisations de ceux-ci

Non-Patent Citations (20)

* Cited by examiner, † Cited by third party
Title
"Approved Standard- Seventh Edition", 2007, CLSI, article "M11-A7 Methods for Antimicrobial Susceptibility Testing of Anaerobic Bacteria"
"Approved Standard-Eighth Edition", 2009, article "M07-A8 Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria that Grow Aerobically"
CHAMPOUX, J.J., ANNU. REV. BIOCHEM., vol. 70, 2001, pages 369 - 413
CHARIFSON ET AL., J. MED. CHEM., vol. 51, 2008, pages 5243 - 5263
DE CLERQ ET AL., CURRENT OPINION IN ANTI-INFECTIVE INVESTIGATIONAL DRUGS, vol. 1, 1999, pages 1
DRLICA, MOLECULAR MICROBIOLOGY, vol. 6, 1992, pages 425
DRLICA; ZHAO, MICROBIOLOGY AND MOLECULAR BIOLOGY REVIEWS, vol. 61, 1997, pages 377 - 392
GERSHMAN, THE MEDICAL REPORTER, 1997
HOFMA ET AL.: "Recent Developments in Coated Stents", CURRENT INTERVENTIONAL CARDIOLOGY REPORTS, vol. 3, 2001, pages 28 - 36
KORNBERG; BAKER: "DNA Replication, 2d Ed.,", 1992, W. H. FREEMAN AND CO.
LEVY: "The Challenge of Antibiotic Resistance", SCIENTIFIC AMERICAN, March 1998 (1998-03-01)
LEWIS, FDA CONSUMER MAGAZINE, September 1995 (1995-09-01)
LEWIS: "The Rise of AntibioticResistant Infections", FDA CONSUMER MAGAZINE, September 1995 (1995-09-01)
MAXWELL, MOL. MICROBIOL., vol. 9, 1993, pages 681
MAXWELL, TRENDS IN MICROBIOLOGY, vol. 5, 1997, pages 102
PAUL S. CHARIFSON ET AL: "Novel Dual-Targeting Benzimidazole Urea Inhibitors of DNA Gyrase and Topoisomerase IV Possessing Potent Antibacterial Activity: Intelligent Design and Evolution through the Judicious Use of Structure-Guided Design and Stucture-Activity Relationships", JOURNAL OF MEDICINAL CHEMISTRY, vol. 51, no. 17, 11 September 2008 (2008-09-11), pages 5243 - 5263, XP055021427, ISSN: 0022-2623, DOI: 10.1021/jm800318d *
TAMURA; GELLERT, J. BIOL. CHEM., vol. 265, 1990, pages 21342
TSUBERY, H. ET AL., J. MED. CHEM., 2000, pages 3085 - 3092
V. LORIAN: "Antibiotics in Laboratory Medicine", 1996, WILLIAMS AND WILKINS
VAARA, M., MICROBIOLOGICAL REVIEWS, 1992, pages 395 - 41 L

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9040542B2 (en) 2011-01-14 2015-05-26 Vertex Pharmaceuticals Incorporated Solid forms of gyrase inhibitor (R)-1-ethyl-3-[6-fluoro-5-[2-(1-hydroxy-1-methyl-ethyl)pryimidin-5-yl]-7-(tetrahydrofuran-2-yl)-1H-benzimidazol-2-yl]urea
US8912326B2 (en) 2011-01-14 2014-12-16 Vertex Pharmaceuticals Incorporated Process of making gyrase and topoisomerase inhibitors
US8969359B2 (en) 2011-01-14 2015-03-03 Vertex Pharmaceuticals Incorporated Gyrase and topoisomerase IV inhibitors
WO2012177707A1 (fr) * 2011-06-20 2012-12-27 Vertex Pharmaceuticals Incorporated Esters phosphoriques d'inhibiteurs de gyrases et de topoisomérases
US9125922B2 (en) 2011-06-20 2015-09-08 Vertex Pharmaceuticals Incorporated Phosphate esters of gyrase and topoisomerase inhibitors
WO2014014845A1 (fr) * 2012-07-18 2014-01-23 Vertex Pharmaceuticals Incorporated Thérapie combinée comprenant de la 1-éthyl-3[5-[-2-{1-hydroxy-1-méthyl-éthyl}pyrimidin-5-yl]-7-(tétrahydrofuran-2-yl}-1h-benzimidazol-2-yl]urée et ses dérivés pour traiter des maladies mycobactériennes
WO2014015105A1 (fr) * 2012-07-18 2014-01-23 Vertex Pharmaceuticals Incorporated Formes solides de (r)-2-(5-(2-(3-éthylureido)-6-fluoro-7-(tétrahydrofuran-2-yl)-1h-benzo[d]imidazol-5-yl)pyrimidin-2-yl)propan-2-yl dihydrogen phosphate et ses sels
US9018216B2 (en) 2012-07-18 2015-04-28 Vertex Pharmaceuticals Incorporated Solid forms of (R)-2-(5-(2-(3-ethylureido)-6-fluoro-7-(tetrahydrofuran-2-yl)-1H-benzo[d]imidazol-5-yl)pyrimidin-2-yl)propan-2-yl dihydrogen phosphate and salts thereof
US9572809B2 (en) 2012-07-18 2017-02-21 Spero Trinem, Inc. Combination therapy to treat Mycobacterium diseases
US9937192B2 (en) 2012-07-18 2018-04-10 Spero Trinem, Inc. Combination therapy to treat mycobacterium diseases
WO2015114452A2 (fr) 2014-02-03 2015-08-06 Biota Europe Ltd Composés antibactériens
WO2018174288A1 (fr) 2017-03-24 2018-09-27 大正製薬株式会社 Dérivé de 2(1h)-quinolinone
KR20190133667A (ko) 2017-03-24 2019-12-03 다이쇼 세이야꾸 가부시끼가이샤 2(1h)-퀴놀리논 유도체

Also Published As

Publication number Publication date
AU2012205415B2 (en) 2017-02-02
TWI546298B (zh) 2016-08-21
RU2609259C2 (ru) 2017-01-31
CA2824516C (fr) 2019-02-26
KR20140037031A (ko) 2014-03-26
AU2012205415A1 (en) 2013-08-01
AR084863A1 (es) 2013-07-10
IL227406A (en) 2017-02-28
TW201309677A (zh) 2013-03-01
RU2013137750A (ru) 2015-02-20
US8481551B2 (en) 2013-07-09
CN103443096B (zh) 2016-06-29
BR112013017974B1 (pt) 2021-05-25
US20130289002A1 (en) 2013-10-31
EP2663557B1 (fr) 2015-05-27
CA2824516A1 (fr) 2012-07-19
NZ612961A (en) 2015-10-30
BR112013017974A2 (pt) 2018-05-02
MX2013008162A (es) 2013-08-27
ES2545516T3 (es) 2015-09-11
JP6085829B2 (ja) 2017-03-01
SG191946A1 (en) 2013-08-30
CL2013002025A1 (es) 2013-12-27
US8969359B2 (en) 2015-03-03
EP2663557A1 (fr) 2013-11-20
US20120184512A1 (en) 2012-07-19
IL227406A0 (en) 2013-09-30
MX339455B (es) 2016-05-27
KR101897952B1 (ko) 2018-09-12
JP2014503557A (ja) 2014-02-13
CN103443096A (zh) 2013-12-11
ZA201305233B (en) 2014-11-26

Similar Documents

Publication Publication Date Title
EP2663557B1 (fr) Inhibiteurs de pyrimidine gyrase et topoïsomérase iv
EP2721026B1 (fr) Esters de phosphate de gyrase et inhibiteurs de la topoisomérase
US9018216B2 (en) Solid forms of (R)-2-(5-(2-(3-ethylureido)-6-fluoro-7-(tetrahydrofuran-2-yl)-1H-benzo[d]imidazol-5-yl)pyrimidin-2-yl)propan-2-yl dihydrogen phosphate and salts thereof
EP2663562B1 (fr) Formes solides d'inhibiteur de gyrase (r)-1-éthyle-3-[6-fluoro-5-[2-(1-hydroxy-1-méthyle-éthyle) pyrimidine-5-yl]-7-(tétrahydrofurane-2-yl)-1h-benzimidazol-2-yl]urée
US8481552B2 (en) Solid forms of gyrase inhibitor (R)-1-ethyl-3-[5-[2-(1-hydroxy-1-methyl-ethyl)pyrimidin-5-YL]-7-(tetrahydrofuran-2-yl)-1H-benzimidazol-2-YL]urea
NZ612961B2 (en) Pyrimidine gyrase and topoisomerase iv inhibitors

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12702920

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2824516

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2013549575

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2012702920

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20137018403

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2013/008162

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2012205415

Country of ref document: AU

Date of ref document: 20120113

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2013137750

Country of ref document: RU

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112013017974

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112013017974

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20130712